Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
Add more filters










Publication year range
1.
Eur J Pharm Biopharm ; 86(3): 514-23, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24361461

ABSTRACT

Gemcitabine prodrugs with D- and L-configuration amino acids were synthesized and their chemical stability in buffers, resistance to glycosidic bond metabolism, enzymatic activation, permeability in Caco-2 cells and mouse intestinal membrane, anti-proliferation activity in cancer cell were determined and compared to that of parent drug, gemcitabine. Prodrugs containing D-configuration amino acids were enzymatically more stable than ones with L-configuration amino acids. The activation of all gemcitabine prodrugs was 1.3-17.6-fold faster in cancer cell homogenate than their hydrolysis in buffer, suggesting enzymatic action. The enzymatic activation of amino acid monoester prodrugs containing D-configuration amino acids in cell homogenates was 2.2-10.9-fold slower than one of amino acid monoester prodrugs with L-configuration amino acids. All prodrugs exhibited enhanced resistance to glycosidic bond metabolism by thymidine phosphorylase compared to parent gemcitabine. Gemcitabine prodrugs showed superior the effective permeability in mouse jejunum to gemcitabine. More importantly, the high plasma concentration of d-amino acid gemcitabine prodrugs was observed more than one of L-amino acid gemcitabine prodrugs. In general, the 5'-mono-amino acid monoester gemcitabine prodrugs exhibited higher permeability and uptake than their parent drug, gemcitabine. Cell proliferation assays in AsPC-1 pancreatic ductal cell line indicated that gemcitabine prodrugs were more potent than their parent drug, gemcitabine. The transport and enzymatic profiles of 5'-D-valyl-gemcitabine and 5'-D-phenylalanyl-gemcitabine suggest their potential for increased oral uptake and delayed enzymatic bioconversion as well as enhanced uptake and cytotoxic activity in cancer cells, would facilitate the development of oral dosage form for anti-cancer agents and, hence, improve the quality of life for the cancer patients.


Subject(s)
Antimetabolites, Antineoplastic/metabolism , Cell Membrane Permeability/physiology , Deoxycytidine/analogs & derivatives , Prodrugs/metabolism , Administration, Oral , Amino Acids/chemistry , Amino Acids/metabolism , Animals , Antimetabolites, Antineoplastic/administration & dosage , Antimetabolites, Antineoplastic/chemistry , Caco-2 Cells , Cell Membrane Permeability/drug effects , Deoxycytidine/administration & dosage , Deoxycytidine/chemistry , Deoxycytidine/metabolism , Enzyme Stability/drug effects , Enzyme Stability/physiology , Female , Humans , Mice , Mice, Inbred BALB C , Prodrugs/administration & dosage , Prodrugs/chemistry , Stereoisomerism , Gemcitabine
2.
Mol Pharm ; 10(2): 445-58, 2013 Feb 04.
Article in English | MEDLINE | ID: mdl-23339402

ABSTRACT

Certain acyclic nucleoside phosphonates (ANPs) such as (S)-HPMPC (cidofovir, Vistide) and (S)-HPMPA have been shown to be active against a broad spectrum of DNA and retroviruses. However, their poor absorption as well as their toxicity limit the utilization of these therapeutics in the clinic. Nucleoside phosphonates are poorly absorbed primarily due to the presence of the phosphonic acid group, which ionizes at physiological pH. When dosed intravenously they display dose-limiting nephrotoxicity due to their accumulation in the kidney. To overcome these limitations, nucleoside phosphonate prodrug strategies have taken center stage in the development pathway and a number of different approaches are at various stages of development. Our efforts have focused on the development of ANP prodrugs in which a benign amino acid promoiety masks a phosphonate P-OH via a hydroxyl side chain. The design of these prodrugs incorporates multiple chemical groups (the P-X-C linkage, the amino acid stereochemistry, the C-terminal and N-terminal functional groups) that can be tuned to modify absorption, pharmacokinetic and efficacy properties with the goal of improving overall prodrug performance.


Subject(s)
Amino Acids/chemistry , Antiviral Agents/chemistry , Cytosine/analogs & derivatives , Organophosphonates/chemistry , Prodrugs/chemistry , Cidofovir , Cytosine/chemistry , Molecular Structure
3.
J Med Chem ; 54(16): 5680-93, 2011 Aug 25.
Article in English | MEDLINE | ID: mdl-21812420

ABSTRACT

Eight novel single amino acid (6-11) and dipeptide (12, 13) tyrosine P-O esters of cyclic cidofovir ((S)-cHPMPC, 4) and its cyclic adenine analogue ((S)-cHPMPA, 3) were synthesized and evaluated as prodrugs. In vitro IC(50) values for the prodrugs (<0.1-50 µM) vs vaccinia, cowpox, human cytomegalovirus, and herpes simplex type 1 virus were compared to those for the parent drugs ((S)-HPMPC, 2; (S)-HPMPA, 1; IC(50) 0.3-35 µM); there was no cytoxicity with KB or HFF cells at ≤100 µM. The prodrugs exhibited a wide range of half-lives in rat intestinal homogenate at pH 6.5 (<30-1732 min) with differences of 3-10× between phostonate diastereomers. The tyrosine alkylamide derivatives of 3 and 4 were the most stable. (l)-Tyr-NH-i-Bu cHPMPA (11) was converted in rat or mouse plasma solely to two active metabolites and had significantly enhanced oral bioavailability vs parent drug 1 in a mouse model (39% vs <5%).


Subject(s)
Adenine/analogs & derivatives , Cytosine/analogs & derivatives , Organophosphonates/chemistry , Prodrugs/chemistry , Tyrosine/chemistry , Adenine/chemistry , Adenine/pharmacokinetics , Adenine/pharmacology , Animals , Antiviral Agents/chemistry , Antiviral Agents/pharmacokinetics , Antiviral Agents/pharmacology , Area Under Curve , Biological Availability , Cell Line, Tumor , Cell Survival/drug effects , Cells, Cultured , Cidofovir , Cowpox virus/drug effects , Cytomegalovirus/drug effects , Cytosine/chemistry , Cytosine/pharmacokinetics , Cytosine/pharmacology , Fibroblasts/cytology , Fibroblasts/drug effects , Herpesvirus 1, Human/genetics , Humans , Inhibitory Concentration 50 , Mice , Models, Chemical , Molecular Structure , Organophosphonates/pharmacokinetics , Organophosphonates/pharmacology , Prodrugs/pharmacokinetics , Prodrugs/pharmacology , Rats , Vaccinia virus/drug effects
4.
Pharm Res ; 28(10): 2575-88, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21671137

ABSTRACT

PURPOSE: To evaluate the advantages of amino acid/dipeptide monoester prodrugs for cancer treatments by assessing the uptake and cytotoxic effects of floxuridine prodrugs in a secondary cancer cell monolayer following permeation across a primary cancer cell monolayer. METHODS: The first Capan-2 monolayer was grown on membrane transwell inserts; the second monolayer was grown at the bottom of a plate. The permeation of floxuridine and its prodrugs across the first monolayer and the uptake and cell proliferation assay on secondary layer were sequentially determined. RESULTS: All floxuridine prodrugs exhibited greater permeation across the first Capan-2 monolayer than the parent drug. The correlation between uptake and growth inhibition in the second monolayer with intact prodrug permeating the first monolayer suggests that permeability and enzymatic stability are essential for sustained action of prodrugs in deeper layers of tumors. The correlation of uptake and growth inhibition were vastly superior for dipeptide prodrugs to those obtained with mono amino acid prodrugs. CONCLUSIONS: Although a tentative general overall correlation between intact prodrug and uptake or cytotoxic action was obtained, it appears that a mixture of floxuridine prodrugs with varying beneficial characteristics may be more effective in treating tumors.


Subject(s)
Amino Acids/administration & dosage , Dipeptides/administration & dosage , Floxuridine/administration & dosage , Prodrugs/administration & dosage , Amino Acids/pharmacokinetics , Cell Line, Tumor , Cell Proliferation/drug effects , Delayed-Action Preparations , Dipeptides/pharmacokinetics , Drug Delivery Systems/methods , Drug Stability , Enzyme Stability/drug effects , Floxuridine/pharmacokinetics , Humans , Neoplasms/drug therapy , Neoplasms/metabolism , Permeability/drug effects , Prodrugs/chemical synthesis , Prodrugs/pharmacokinetics
5.
Mol Pharm ; 7(5): 1827-34, 2010 Oct 04.
Article in English | MEDLINE | ID: mdl-20701326

ABSTRACT

The FDA classifies a drug substance as high-permeability when the fraction of dose absorbed (F(abs)) in humans is 90% or higher. This direct correlation between human permeability and F(abs) has been recently controversial, since the ß-blocker sotalol showed high F(abs) (90%) and low Caco-2 permeability. The purpose of this study was to investigate the scientific basis for this disparity between permeability and F(abs). The effective permeabilities (P(eff)) of sotalol and metoprolol, a FDA standard for the low/high P(eff) class boundary, were investigated in the rat perfusion model, in three different intestinal segments with pHs corresponding to the physiological pH in each region: (1) proximal jejunum, pH 6.5; (2) mid small intestine, pH 7.0; and (3) distal ileum, pH 7.5. Both metoprolol and sotalol showed pH-dependent permeability, with higher P(eff) at higher pH. At any given pH, sotalol showed lower permeability than metoprolol; however, the permeability of sotalol determined at pH 7.5 exceeded/matched metoprolol's at pH 6.5 and 7.0, respectively. Physicochemical analysis based on ionization, pK(a) and partitioning of these drugs predicted the same trend and clarified the mechanism behind these observed results. Experimental octanol-buffer partitioning experiments confirmed the theoretical curves. An oral dose of metoprolol has been reported to be completely absorbed in the upper small intestine; it follows, hence, that metoprolol's P(eff) value at pH 7.5 is not likely physiologically relevant for an immediate release dosage form, and the permeability at pH 6.5 represents the actual relevant value for the low/high permeability class boundary. Although sotalol's permeability is low at pH 6.5 and 7.0, at pH 7.5 it exceeds/matches the threshold of metoprolol at pH 6.5 and 7.0, most likely responsible for its high F(abs). In conclusion, we have shown that, in fact, there is no discrepancy between P(eff) and F(abs) in sotalol's absorption; the data emphasize that, if a compound has high fraction of dose absorbed, it will have high-permeability, not necessarily in the jejunum, but at some point along the relevant intestinal regions.


Subject(s)
Chemistry, Pharmaceutical , Pharmacokinetics , Animals , Caco-2 Cells , Drug Discovery , Humans , Hydrogen-Ion Concentration , Intestinal Absorption , Male , Metoprolol/administration & dosage , Metoprolol/pharmacokinetics , Permeability , Rats , Rats, Wistar , Sotalol/administration & dosage , Sotalol/pharmacokinetics , Therapeutic Equivalency , United States , United States Food and Drug Administration
6.
Antiviral Res ; 85(3): 482-9, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19969024

ABSTRACT

Cidofovir (HPMPC) is a broad-spectrum antiviral agent, currently used to treat AIDS-related human cytomegalovirus retinitis. Cidofovir has recognized therapeutic potential for orthopox virus infections, although its use is hampered by its inherent low oral bioavailability. Val-Ser-cyclic HPMPC (Val-Ser-cHPMPC) is a promising peptide prodrug which has previously been shown by us to improve the permeability and bioavailability of the parent compound in rodent models (Eriksson et al., 2008. Molecular Pharmaceutics 5, 598-609). Puromycin-sensitive aminopeptidase was partially purified from Caco-2 cell homogenates and identified as a prodrug activating enzyme for Val-Ser-cHPMPC. The prodrug activation process initially involves an enzymatic step where the l-Valine residue is removed by puromycin-sensitive aminopeptidase, a step that is bestatin-sensitive. Subsequent chemical hydrolysis results in the generation of cHPMPC. A recombinant puromycin-sensitive aminopeptidase was generated and its substrate specificity investigated. The k(cat) for Val-pNA was significantly lower than that for Ala-pNA, suggesting that some amino acids are preferred over others. Furthermore, the three-fold higher k(cat) for Val-Ser-cHPMPC as compared to Val-pNA suggests that the leaving group may play an important role in determining hydrolytic activity. In addition to its ability to hydrolyze a variety of substrates, these observations strongly suggest that puromycin-sensitive aminopeptidase is an important enzyme for activating Val-Ser-cHPMPC in vivo. Taken together, our data suggest that puromycin-sensitive aminopeptidase makes an attractive target for future prodrug design.


Subject(s)
Aminopeptidases/metabolism , Antiviral Agents/metabolism , Cytosine/analogs & derivatives , Organophosphonates/metabolism , Prodrugs/metabolism , Aminopeptidases/genetics , Aminopeptidases/isolation & purification , Caco-2 Cells , Cidofovir , Cytosine/metabolism , Humans , Kinetics , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Substrate Specificity
8.
Molecules ; 13(7): 1441-54, 2008 Jun 28.
Article in English | MEDLINE | ID: mdl-18719516

ABSTRACT

A series of amino acid monoester prodrugs of floxuridine was synthesized and evaluated for the improvement of oral bioavailability and the feasibility of target drug delivery via oligopeptide transporters. All floxuridine 5'-amino acid monoester prodrugs exhibited PEPT1 affinity, with inhibition coefficients of Gly-Sar uptake (IC50) ranging from 0.7 - 2.3 mM in Caco-2 and 2.0 - 4.8 mM in AsPC-1 cells, while that of floxuridine was 7.3 mM and 6.3 mM, respectively. Caco-2 membrane permeabilities of floxuridine prodrugs (1.01 - 5.31 x 10(-6 )cm/sec) and floxuridine (0.48 x 10(-6 )cm/sec) were much higher than that of 5-FU (0.038 x 10(-6) cm/sec). MDCK cells stably transfected with the human oligopeptide transporter PEPT1 (MDCK/hPEPT1) exhibited enhanced cell growth inhibition in the presence of the prodrugs. This prodrug strategy offers great potential, not only for increased drug absorption but also for improved tumor selectivity and drug efficacy.


Subject(s)
Amino Acids/pharmacology , Antimetabolites, Antineoplastic/pharmacology , Cell Proliferation/drug effects , Floxuridine/pharmacology , Floxuridine/pharmacokinetics , Prodrugs/pharmacology , Prodrugs/pharmacokinetics , Symporters/metabolism , Absorption/drug effects , Amino Acids/chemical synthesis , Amino Acids/pharmacokinetics , Animals , Antimetabolites, Antineoplastic/chemical synthesis , Caco-2 Cells , Dogs , Drug Delivery Systems , Esters , Growth Inhibitors/pharmacokinetics , Growth Inhibitors/pharmacology , Humans , Peptide Transporter 1 , Prodrugs/chemical synthesis , Symporters/biosynthesis , Symporters/genetics
9.
Mol Pharm ; 5(5): 717-27, 2008.
Article in English | MEDLINE | ID: mdl-18652477

ABSTRACT

Dipeptide monoester prodrugs of floxuridine were synthesized, and their chemical stability in buffers, resistance to glycosidic bond metabolism, affinity for PEPT1, enzymatic activation and permeability in cancer cells were determined and compared to those of mono amino acid monoester floxuridine prodrugs. Prodrugs containing glycyl moieties were the least stable in pH 7.4 buffer ( t 1/2 < 100 min). The activation of all floxuridine prodrugs was 2- to 30-fold faster in cell homogenates than their hydrolysis in buffer, suggesting enzymatic action. The enzymatic activation of dipeptide monoester prodrugs containing aromatic promoieties in cell homogenates was 5- to 20-fold slower than that of other dipeptide and most mono amino acid monoester prodrugs ( t 1/2 approximately 40 to 100 min). All prodrugs exhibited enhanced resistance to glycosidic bond metabolism by thymidine phosphorylase compared to parent floxuridine. In general, the 5'-O-dipeptide monoester floxuridine prodrugs exhibited higher affinity for PEPT1 than the corresponding 5'-O-mono amino acid ester prodrugs. The permeability of dipeptide monoester prodrugs across Caco-2 and Capan-2 monolayers was 2- to 4-fold higher than the corresponding mono amino acid ester prodrug. Cell proliferation assays in AsPC-1 and Capan-2 pancreatic ductal cell lines indicated that the dipeptide monoester prodrugs were equally as potent as mono amino acid prodrugs. The transport and enzymatic profiles of 5'- l-phenylalanyl- l-tyrosyl-floxuridine, 5'- l-phenylalanyl- l-glycyl-floxuridine, and 5'- l-isoleucyl- l-glycyl-floxuridine suggest their potential for increased oral uptake, delayed enzymatic bioconversion and enhanced resistance to metabolism to 5-fluorouracil, as well as enhanced uptake and cytotoxic activity in cancer cells, attributes that would facilitate prolonged systemic circulation for enhanced therapeutic action.


Subject(s)
Antineoplastic Agents/pharmacology , Colonic Neoplasms/drug therapy , Dipeptides/chemistry , Floxuridine/analogs & derivatives , Floxuridine/chemistry , Pancreatic Neoplasms/drug therapy , Prodrugs/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Biological Transport , Cell Line, Tumor , Cell Proliferation/drug effects , Dipeptides/metabolism , Dipeptides/pharmacology , Drug Screening Assays, Antitumor , Drug Stability , Floxuridine/metabolism , Floxuridine/pharmacology , Humans , Molecular Structure , Prodrugs/chemistry , Prodrugs/metabolism , Thymidine Phosphorylase/chemistry
10.
Mol Pharm ; 5(4): 598-609, 2008.
Article in English | MEDLINE | ID: mdl-18481868

ABSTRACT

Cidofovir (HPMPC, 1), a broad-spectrum antiviral agent, is currently used to treat AIDS-related human cytomegalovirus (HCMV) retinitis and has recognized therapeutic potential for orthopox virus infections, but is limited by its low oral bioavailability. Cyclic cidofovir (2) displays decreased nephrotoxicity compared to 1, while also exhibiting potent antiviral activity. Here we describe in detail the synthesis and evaluation as prodrugs of four cHPMPC dipeptide conjugates in which the free POH of 2 is esterified by the Ser side chain alcohol group of an X-L-Ser(OMe) dipeptide: 3 (X=L-Ala), 4 (X=L-Val), 5 (X=L-Leu), and 6 (X=L-Phe). Perfusion studies in the rat establish that the mesenteric permeability to 4 is more than 20-fold greater than to 1, and the bioavailability of 4 is increased 6-fold relative to 1 in an in vivo murine model. In gastrointestinal and liver homogenates, the cHPMPC prodrugs are rapidly hydrolyzed to 2. Prodrugs 3, 4, and 5 are nontoxic at 100 microM in HFF and KB cells and in cell-based plaque reduction assays had IC 50 values of 0.1-0.5 microM for HCMV and 10 microM for two orthopox viruses (vaccinia and cowpox). The enhanced transport properties of 3-6, conferred by incorporation of a biologically benign dipeptide moiety, and the facile cleavage of the Ser-O-P linkage suggest that these prodrugs represent a promising new approach to enhancing the bioavailability of 2.


Subject(s)
Antiviral Agents/chemical synthesis , Antiviral Agents/pharmacology , Cytosine/analogs & derivatives , Organophosphonates/chemical synthesis , Organophosphonates/pharmacology , Peptides/chemistry , Prodrugs/chemical synthesis , Prodrugs/pharmacology , Animals , Antiviral Agents/blood , Antiviral Agents/chemistry , Cell Line, Tumor , Cidofovir , Cytomegalovirus/drug effects , Cytosine/blood , Cytosine/chemical synthesis , Cytosine/chemistry , Cytosine/pharmacology , Esterification , Humans , Hydrolysis , Intestinal Mucosa/metabolism , Male , Mice , Models, Molecular , Molecular Structure , Organophosphonates/blood , Organophosphonates/chemistry , Prodrugs/chemistry , Prodrugs/metabolism , Rats , Serine/chemistry , Static Electricity
11.
Pharm Res ; 25(7): 1591-600, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18213452

ABSTRACT

PURPOSE: The study was designed to evaluate the effect of delayed release (DR) on absorption and bioavailability of intestinally metabolized drugs after oral dosing, using the HMG-CoA reductase inhibitor simvastatin, a CYP3A substrate, as a model drug. MATERIALS AND METHODS: To target drug release and to assess regional gastrointestinal absorption of the CYP 3A substrate simvastatin from the distal parts of the intestine, delayed release film coated tableted oral dosage forms were developed. Simvastatin delayed release tablet, simvastatin immediate release capsule and simvastatin immediate release tablet Zocor were administered as single doses (20 mg) to fasting healthy volunteers in a crossover design. RESULTS: Simvastatin bioavailability was increased by a factor of three, as compared to the reference formulation Zocor. The overall metabolite levels from the immediate release capsules tended to be higher throughout the period studied than the metabolite levels following administration of Zocor and simvastatin delayed release dosage form. CONCLUSIONS: The interplay between gastrointestinal physiology (lower CYP 3A expression in the distal ileum and the colon) and formulation design (zero-order controlled release after a predetermined lag-time) resulted in successful absorption and bioavailability improvement and represent a viable strategy to reduce the dose of CYP 3A drugs.


Subject(s)
Cytochrome P-450 CYP3A/metabolism , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacokinetics , Simvastatin/pharmacokinetics , Acrylic Resins , Adult , Algorithms , Area Under Curve , Biological Availability , Capsules , Chromatography, High Pressure Liquid , Cross-Over Studies , Delayed-Action Preparations , Excipients , Female , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , Intestinal Absorption , Male , Simvastatin/administration & dosage , Solubility , Spectrophotometry, Ultraviolet
12.
Eur J Pharm Biopharm ; 67(3): 752-8, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17560100

ABSTRACT

The therapeutic efficacy of prophalan-L, the L-proline prodrug of melphalan that demonstrated prolidase-dependent bioactivation to melphalan, was examined in vivo in a mouse melanoma model. Prophalan-L exhibited 2- to 2.5-fold higher hydrolytic and cytotoxic activity than prophalan-D, the D-analog, in B16-F10 murine melanoma cells in vitro. Prophalan-L cytotoxicity in B16-F10 cells was lower (GI50=221 microM) than that of melphalan (GI50=173 microM). The tumor growth profiles in C57BL/6J mice injected with B16-F10 cells and treated with melphalan (5.5 microg/g i.p.) and equimolar concentrations of the prodrugs demonstrated significant difference between the control (buffered saline) and melphalan or prophalan-L but no significant difference between control and prophalan-D or between melphalan and prophalan-L. Prophalan-L was significantly less toxic than melphalan, while no significant difference was observed in toxicity, measured as percent weight loss, between the prodrugs and saline control. Tumor reduction efficacy at high doses (12 microg/g i.p.) was similar for melphalan and prophalan-L; however, fatal toxicity was associated with melphalan while prophalan-L exhibited significantly lower systemic toxicity. An excellent correlation between GI50 and tumor reduction efficacy was observed for the tested drugs (r2=0.95). Prophalan-L thus demonstrates higher therapeutic index than melphalan in the murine melanoma model.


Subject(s)
Antineoplastic Agents, Alkylating/therapeutic use , Melanoma, Experimental/drug therapy , Melphalan/analogs & derivatives , Melphalan/therapeutic use , Prodrugs/therapeutic use , Proline/analogs & derivatives , Animals , Antineoplastic Agents, Alkylating/metabolism , Body Weight/drug effects , Calorimetry , Cell Proliferation/drug effects , Chromatography, High Pressure Liquid , Female , Hydrolysis , Indicators and Reagents , Melanoma, Experimental/pathology , Melphalan/metabolism , Mice , Mice, Inbred C57BL , Prodrugs/metabolism , Proline/metabolism , Proline/therapeutic use , Spectrophotometry, Ultraviolet , Stereoisomerism , Survival Analysis
13.
Mol Pharm ; 4(1): 95-103, 2007.
Article in English | MEDLINE | ID: mdl-17274667

ABSTRACT

Bile acids conjugated to oligoarginine-containing peptides (BACs) form complexes with DNA based on the electrostatic interactions between negatively charged phosphate groups of the nucleic acid and the positively charged side chain guanidinium groups of the oligoarginine in the BACs. Charge neutralization of both components and subsequent increases of the net positive charge of the complex combined with the water-soluble lipophilic nature of the bile acid results in changes in the physicochemistry and biological properties of the complexes. We have examined the relationship of a series of 13 BACs on their interaction with circular plasmid DNA (pDNA). The formation of soluble, low-density and insoluble, high-density complexes was analyzed using several methods. The formation of high-density complexes was dependent on the DNA concentration, and was enhanced by increasing the BAC to pDNA charge ratio. Several of the BAC:pDNA complexes demonstrated exclusion of the DNA-intercalator Hoechst 33258 from pDNA, and were also protected from DNase activity. Several BAC conjugates interacted with pDNA to form nanometer-sized particles suitable for cell transfection in vitro. Five of the 13 BACs were transfection competent as single agents, and 11 of the 13 BACs showed enhancement of transfection in combination with DOPE containing liposomes or silica nanoparticles.


Subject(s)
Bile Acids and Salts/metabolism , DNA, Circular/metabolism , Oligopeptides/metabolism , Transfection/methods , Amino Acid Sequence , Animals , Bile Acids and Salts/chemistry , Bisbenzimidazole/metabolism , DNA, Circular/ultrastructure , Deoxyribonucleases/metabolism , Fluorescent Dyes/metabolism , Humans , Liposomes/metabolism , Mice , Micelles , Microscopy, Electron, Transmission , Molecular Sequence Data , NIH 3T3 Cells , Nanoparticles , Nephelometry and Turbidimetry , Oligopeptides/chemical synthesis , Oligopeptides/chemistry , Particle Size , Plasmids/metabolism , Protein Binding
14.
Bioorg Med Chem Lett ; 17(3): 583-6, 2007 Feb 01.
Article in English | MEDLINE | ID: mdl-17161946

ABSTRACT

Cidofovir (HPMPC) is a broad-spectrum anti-viral agent whose potential, particularly in biodefense scenarios, is limited by its low oral bioavailability. Two prodrugs (3 and 4) created by conjugating ethylene glycol-linked amino acids (L-Val, L-Phe) with the cyclic form of cidofovir (cHPMPC) via a P-O ester bond were synthesized and their pH-dependent stability (3 and 4), potential for in vivo reconversion to drug (3), and oral bioavailability (3) were evaluated. The prodrugs were stable in buffer between pH 3 and 5, but underwent rapid hydrolysis in liver (t(1/2) = 3.7 min), intestinal (t(1/2) = 12.5 min), and Caco-2 cell homogenates (t(1/2) = 20.2 min). In vivo (rat), prodrug 3 was >90% reconverted to cHPMPC. The prodrug was 4x more active than ganciclovir (IC50 value, 0.68 microM vs 3.0 microM) in a HCMV plaque reduction assay. However, its oral bioavailability in a rat model was similar to the parent drug. The contrast between the promising activation properties and unenhanced transport of the prodrug is briefly discussed.


Subject(s)
Amino Acids/chemistry , Antiviral Agents/chemical synthesis , Antiviral Agents/metabolism , Cytosine/analogs & derivatives , Ethylene Glycol/chemistry , Organophosphonates/chemical synthesis , Organophosphonates/metabolism , Prodrugs/chemical synthesis , Prodrugs/metabolism , Animals , Antiviral Agents/pharmacokinetics , Biological Availability , Biotransformation , Cell Survival/drug effects , Cidofovir , Cytosine/chemical synthesis , Cytosine/metabolism , Cytosine/pharmacokinetics , Half-Life , Humans , Hydrolysis , KB Cells , Magnetic Resonance Spectroscopy , Organophosphonates/pharmacokinetics , Peptide Transporter 1 , Phenylalanine/chemistry , Rats , Symporters/metabolism , Valine/chemistry , Viral Plaque Assay
15.
Pharm Res ; 22(9): 1510-8, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16132363

ABSTRACT

PURPOSE: The aim of this study was to synthesize amino acid ester prodrugs of 5-fluoro-2'-deoxyuridine (floxuridine) to enhance intestinal absorption and resistance to glycosidic bond metabolism. METHODS: Amino acid ester prodrugs were synthesized and examined for their hydrolytic stability in human plasma, in Caco-2 cell homogenates, and in the presence of thymidine phosphorylase. Glycyl-L: -sarcosine uptake inhibition and direct uptake studies with HeLa/PEPT1 cells [HeLa cells overexpressing oligopeptide transporter (PEPT1)] were conducted to determine PEPT1-mediated transport and compared with permeability of the prodrugs across Caco-2 monolayers. RESULTS: Isoleucyl prodrugs exhibited the highest chemical and enzymatic stability. The prodrugs enhanced the stability of the glycosidic bond of floxuridine. Thymidine phosphorylase rapidly cleaved floxuridine to 5-fluorouracil, whereas with the prodrugs no detectable glycosidic bond cleavage was observed. The 5'-L: -isoleucyl and 5'-L: -valyl monoester prodrugs exhibited 8- and 19-fold PEPT1-mediated uptake enhancement in HeLa/PEPT1 cells, respectively. Uptake enhancement in HeLa/PEPT1 cells correlated highly with Caco-2 permeability for all prodrugs tested. Caco-2 permeability of 5'-L: -isoleucyl and 5'-L: -valyl prodrugs was 8- to 11-fold greater compared with floxuridine. CONCLUSIONS: Amino acid ester prodrugs such as isoleucyl floxuridine that exhibit enhanced Caco-2 transport and slower rate of enzymatic activation to parent, and that are highly resistant to metabolism by thymidine phosphorylase may improve oral delivery and therapeutic index of floxuridine.


Subject(s)
Amino Acids/chemistry , Floxuridine/pharmacology , Glycosides/chemistry , Prodrugs/pharmacology , Biological Availability , Caco-2 Cells , Cell Membrane Permeability , Esterification , Floxuridine/chemistry , Floxuridine/pharmacokinetics , HeLa Cells , Humans , Magnetic Resonance Spectroscopy , Prodrugs/chemistry , Thymidine Phosphorylase/metabolism
16.
J Pharmacol Exp Ther ; 314(2): 883-90, 2005 Aug.
Article in English | MEDLINE | ID: mdl-15901797

ABSTRACT

2-Bromo-5,6-dichloro-1-(beta-d-ribofuranosyl)benzimidazole (BDCRB) is a potent and selective inhibitor of human cytomegalovirus (HCMV), but it lacks clinical utility due to rapid in vivo metabolism. We hypothesized that amino acid ester prodrugs of BDCRB may enhance both in vitro potency and systemic exposure of BDCRB through evasion of BDCRB-metabolizing enzymes. To this end, eight different amino acid prodrugs of BDCRB were tested for N-glycosidic bond stability, ester bond stability, Caco-2 cell uptake, antiviral activity, and cytotoxicity. The prodrugs were resistant to metabolism by BDCRB-metabolizing enzymes, and ester bond cleavage was rate-limiting in metabolite formation from prodrug. Thus, BDCRB metabolism could be controlled by the selection of promoiety. In HCMV plaque-formation assays, l-Asp-BDCRB exhibited 3-fold greater selectivity than BDCRB for inhibition of HCMV replication. This potent and selective antiviral activity in addition to favorable stability profile made l-Asp-BDCRB an excellent candidate for in vivo assessment and pharmacokinetic comparison with BDCRB. In addition to rapid absorption and sufficient prodrug activation after oral administration to mice, l-Asp-BDCRB exhibited a 5-fold greater half-life than BDCRB. Furthermore, the sum of area under the concentration-time profile (AUC)(BDCRB) and AUC(prodrug) after l-Asp-BDCRB administration was roughly 3-fold greater than AUC(BDCRB) after BDCRB administration, suggesting that a reservoir of prodrug was delivered in addition to parent drug. Overall, these findings demonstrate that amino acid prodrugs of BDCRB exhibit evasion of metabolizing enzymes (i.e., bioevasion) in vitro and provide a modular approach for translating this in vitro stability into enhanced in vivo delivery of BDCRB.


Subject(s)
Antiviral Agents/pharmacology , Benzimidazoles/pharmacology , Cytomegalovirus/drug effects , Prodrugs/pharmacology , Ribonucleosides/pharmacology , Animals , Antiviral Agents/pharmacokinetics , Benzimidazoles/pharmacokinetics , Caco-2 Cells , Cell Survival/drug effects , Chromatography, High Pressure Liquid , DNA Glycosylases/metabolism , Female , Humans , Intestinal Mucosa/metabolism , Liver/metabolism , Mice , Prodrugs/pharmacokinetics , Ribonucleosides/pharmacokinetics
17.
Mol Pharm ; 2(1): 37-46, 2005.
Article in English | MEDLINE | ID: mdl-15804176

ABSTRACT

Bioinformatics tools such as Perl, Visual Basic, Cluster, and TreeView were used to analyze public gene expression databases in order to identify potential enzyme targets for prodrug strategies. The analyses indicated that prolidase might be a desirable enzyme target based on its differential expression in melanoma cancer cell lines and its high substrate specificity for dipeptides containing proline at the carboxy terminus. RT-PCR expression of prolidase and hydrolytic activity against N-glycyl-l-proline (GLY-PRO), a standard substrate of prolidase, determined in tumor cell lines, exhibited a high correlation (r(2) = 0.95). These results suggest the possibility of targeting prolidase with prodrugs of anticancer agents for enhanced selectivity. The feasibility of such a scenario was tested by (a) synthesizing prodrugs of melphalan that comprised linkage of the carboxy terminus of the l-phenylalanine moiety of melphalan to the N-terminus of l and d stereoisomers of proline and (b) determining their bioconversion and antiproliferative activities in SK-MEL-5 cells, a melanoma cancer cell line with high expression levels of prolidase. The results of hydrolysis studies of the l- and d-proline prodrugs of melphalan, designated as prophalan-l and prophalan-d, respectively, indicated a approximately 7-fold higher rate of activation of prophalan-l compared to prophalan-d in SK-MEL-5 cell homogenates. Prophalan-l exhibited cytotoxicity (GI(50) = 74.8 microM) comparable to that of melphalan (GI(50) = 57.0 microM) in SK-MEL-5 cells while prophalan-d was ineffective, suggesting that prolidase-specific activation to the parent drug may be essential for cytotoxic action. Thus, melphalan prodrugs such as prophalan-l that are cleavable by prolidase offer the potential for enhanced selectivity by facilitating cytotoxic activity only in cells overexpressing prolidase.


Subject(s)
Dipeptidases/therapeutic use , Drug Design , Melanoma/drug therapy , Melphalan/analogs & derivatives , Prodrugs/therapeutic use , Proline/analogs & derivatives , Proline/chemistry , Amino Acid Sequence , Antineoplastic Agents , Caco-2 Cells , Cell Line, Tumor , Cell Proliferation/drug effects , Colorimetry , Feasibility Studies , Humans , K562 Cells , Melphalan/chemical synthesis , Melphalan/chemistry , Melphalan/metabolism , Melphalan/therapeutic use , Oligonucleotide Array Sequence Analysis , Prodrugs/chemical synthesis , Prodrugs/chemistry , Prodrugs/metabolism , Proline/chemical synthesis , Proline/metabolism , Proline/therapeutic use , Reverse Transcriptase Polymerase Chain Reaction , Substrate Specificity
18.
Mol Pharm ; 2(2): 157-67, 2005.
Article in English | MEDLINE | ID: mdl-15804190

ABSTRACT

Gemcitabine, a clinically effective nucleoside anticancer agent, is a polar drug with low membrane permeability and is administered intravenously. Further, extensive degradation of gemcitabine by cytidine deaminase to an inactive metabolite in the liver affects its activity adversely. Thus, strategies that provide both enhanced transport and high metabolic bioevasion would potentially lead to oral alternatives that may be clinically useful. The objective of this study was to evaluate whether amino acid ester prodrugs of gemcitabine would (a) facilitate transport across intestinal membranes or across cells that express hPEPT1 and (b) provide resistance to deamination by cytidine deaminase. 3'-Monoester, 5'-monoester, and 3',5'-diester prodrugs of gemcitabine utilizing aliphatic (L-valine, D-valine, and L-isoleucine) and aromatic (L-phenylalanine and D-phenylalanine) amino acids as promoieties were synthesized and evaluated for their affinity and direct hPEPT1-mediated transport in HeLa/hPEPT1 cells. All prodrugs exhibited enhanced affinity (IC(50): 0.14-0.16 mM) for the transporter. However, only the 5'-L-valyl and 5'-L-isoleucyl monoester prodrugs exhibited (a) increased uptake (11.25- and 5.64-fold, respectively) in HeLa/hPEPT1 cells compared to HeLa cells and (b) chemical stability in buffers, that were comparable to valacyclovir, a commercially marketed oral amino acid ester prodrug. The widely disparate enzymatic bioconversion profiles of the 5'-L-valyl and 5'-L-isoleucyl prodrugs in Caco-2 cell homogenates along with their significant resistance to deamination by cytidine deaminase suggest that the disposition of gemcitabine following oral administration would be controlled by the rate of bioconversion following transport across the intestinal epithelial membrane. The combined results also suggest that it may be possible to modulate these characteristics by the choice of the amino acid promoiety.


Subject(s)
Amino Acids/pharmacology , Antineoplastic Agents/pharmacology , Deoxycytidine/analogs & derivatives , Prodrugs/chemistry , Symporters/chemistry , Amino Acids/chemistry , Antineoplastic Agents/chemistry , Biological Transport , Caco-2 Cells , Cell Line, Tumor , Chromatography, High Pressure Liquid , Cytidine Deaminase/chemistry , Cytidine Deaminase/metabolism , Deoxycytidine/pharmacokinetics , Deoxycytidine/pharmacology , Dipeptides/pharmacokinetics , Drug Screening Assays, Antitumor/methods , HeLa Cells , Humans , Hydrolysis , Inhibitory Concentration 50 , Peptide Transporter 1 , Peptides/chemistry , Gemcitabine
19.
J Nutr Biochem ; 15(5): 289-95, 2004 May.
Article in English | MEDLINE | ID: mdl-15135153

ABSTRACT

Clinical studies have demonstrated that consumption of phytosterol esters in lipid-based foods decreases serum concentrations of total and LDL cholesterol. These substances represent minimal potential for adverse effects when consumed orally because of their low bioavailability. However, some studies have reported estrogenic and other effects in laboratory animals treated parenterally with phytosterols, demonstrating that these substances may have the potential to cause adverse effects if absorbed. Water-soluble phytosterols have been prepared by formulation with emulsifiers to expand delivery options to include non-lipid-based foods. However, emulsifiers are used as excipients in the formulation of lipophilic pharmaceuticals to increase solubility, thereby increasing their absorption. Therefore, oral consumption of emulsified water-soluble phytosterols could potentially increase their absorption. In the current study, absorption of phytosterols prepared as water-soluble emulsified micelles with two different food-grade emulsifiers was evaluated in Sprague-Dawley rats and compared with absorption of non-micellar free phytosterols and esterified phytosterol mixtures dissolved in a lipophilic vehicle (soybean oil). Rats were dosed via gavage with 42 mg/kg of formulated phytosterol preparations. Blood was collected at 8, 16, 24, and 32 hours, extracted with hexane, derivatized with benzoyl chloride, and analyzed by high-performance liquid chromatography to determine concentrations of beta-sitosterol, and campesterol. Plasma concentrations and AUC(0-32 hours) [microg/mL/h] of beta-sitosterol and campesterol were lower in plasma obtained from rats treated with emulsified phytosterol preparations than in animals treated with free phytosterols dissolved in soybean oil. Because the pharmacokinetic profile of water-soluble phytosterols is similar to that of phytosterols administered in a lipid vehicle, the safety profile is likely to be the same as that of phytosterols and phytosterol esters in currently used applications.


Subject(s)
Cholesterol/analogs & derivatives , Emulsions/pharmacokinetics , Mouth/metabolism , Phytosterols/pharmacokinetics , Absorption , Animals , Cholesterol/blood , Chromatography, High Pressure Liquid , Diet , Esterification , Kinetics , Micelles , Phosphatidylcholines , Phytosterols/administration & dosage , Phytosterols/blood , Rats , Rats, Sprague-Dawley , Sitosterols/blood , Solubility , Water
20.
Pharm Res ; 20(9): 1381-8, 2003 Sep.
Article in English | MEDLINE | ID: mdl-14567631

ABSTRACT

PURPOSE: To synthesize amino acid ester prodrugs of floxuridine (FUdR) and to investigate the effects of structure, stereochemistry, and site of esterification of promoiety on the rates of hydrolysis of these prodrugs in Caco-2 cell homogenates. METHODS: Amino acid ester prodrugs of FUdR were synthesized using established procedures. The kinetics of hydrolysis of prodrugs was evaluated in human adenocarcinoma cell line (Caco-2) homogenates and pH 7.4 phosphate buffer. RESULTS: 3'-Monoester, 5'-monoester, and 3',5'-diester prodrugs of FUdR utilizing proline, L-valine, D-valine, L-phenylalanine, and D-phenylalanine as promoieties were synthesized and characterized. In Caco-2 cell homogenates, the L-amino acid ester prodrugs hydrolyzed 10 to 75 times faster than the corresponding D-amino acid ester prodrugs. Pro and Phe ester prodrugs hydrolyzed much faster (3- to 30-fold) than the corresponding Val ester prodrugs. Further, the 5'-monoester prodrugs hydrolyzed significantly faster (3-fold) than the 3',5'-diester prodrugs. CONCLUSIONS: Novel amino acid ester prodrugs of FUdR were successfully synthesized. The results presented here clearly demonstrate that the rate of FUdR prodrug activation in Caco-2 cell homogenates is affected by the structure, stereochemistry, and site of esterification of the promoiety. Finally, the 5'-Val and 5'-Phe monoesters exhibited desirable characteristics such as good solution stability and relatively fast enzymatic conversion rates.


Subject(s)
Amino Acids/chemistry , Antineoplastic Agents/chemical synthesis , Floxuridine/chemistry , Prodrugs/chemical synthesis , Amino Acids/metabolism , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Buffers , Caco-2 Cells , Chromatography, High Pressure Liquid , Esters , Floxuridine/metabolism , Humans , Hydrolysis , Molecular Structure , Prodrugs/chemistry , Prodrugs/metabolism , Stereoisomerism , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...